80 research outputs found

    Transcriptome analysis of bone marrow mesenchymal stromal cells from patients with primary myelofibrosis

    Get PDF
    International audiencePrimary myelofibrosis (PMF) is a clonal myeloproliferative neoplasm whose severity and treatment complexity are attributed to the presence of bone marrow (BM) fibrosis and alterations of stroma impairing the production of normal blood cells. Despite the recently discovered mutations including the JAK2V617F mutation in about half of patients, the primitive event responsible for the clonal proliferation is still unknown. In the highly inflammatory context of PMF, the presence of fibrosis associated with a neoangiogenesis and an osteosclerosis concomitant to the myeloproliferation and to the increase number of circulating hematopoietic progenitors suggests that the crosstalk between hematopoietic and stromal cells is deregulated in the PMF BM microenvironmental niches. Within these niches, mesenchymal stromal cells (BM-MSC) play a hematopoietic supportive role in the production of growth factors and extracellular matrix which regulate the proliferation, differentiation, adhesion and migration of hematopoietic stem/progenitor cells. A transcriptome analysis of BM-MSC in PMF patients will help to characterize their molecular alterations and to understand their involvement in the hematopoietic stem/progenitor cell deregulation that features PMF

    Tetraspanin CD9 participates in dysmegakaryopoiesis and stromal interactions in primary myelofibrosis

    Get PDF
    Primary myelofibrosis is characterized by clonal myeloproliferation, dysmegakaryopoiesis, extramedullary hematopoiesis associated with myelofibrosis and altered stroma in the bone marrow and spleen. The expression of CD9, a tetraspanin known to participate in megakaryopoiesis, platelet formation, cell migration and interaction with stroma, is deregulated in patients with primary myelofibrosis and is correlated with stage of myelofibrosis. We investigated whether CD9 participates in the dysmegakaryopoiesis observed in patients and whether it is involved in the altered interplay between megakaryocytes and stromal cells. We found that CD9 expression was modulated during megakaryocyte differentiation in primary myelofibrosis and that cell surface CD9 engagement by antibody ligation improved the dysmegakaryopoiesis by restoring the balance of MAPK and PI3K signaling. When co-cultured on bone marrow mesenchymal stromal cells from patients, megakaryocytes from patients with primary myelofibrosis displayed modified behaviors in terms of adhesion, cell survival and proliferation as compared to megakaryocytes from healthy donors. These modifications were reversed after antibody ligation of cell surface CD9, suggesting the participation of CD9 in the abnormal interplay between primary myelofibrosis megakaryocytes and stroma. Furthermore, silencing of CD9 reduced CXCL12 and CXCR4 expression in primary myelofibrosis megakaryocytes as well as their CXCL12-dependent migration. Collectively, our results indicate that CD9 plays a role in the dysmegakaryopoiesis that occurs in primary myelofibrosis and affects interactions between megakaryocytes and bone marrow stromal cells. These results strengthen the “bad seed in bad soil” hypothesis that we have previously proposed, in which alterations of reciprocal interactions between hematopoietic and stromal cells participate in the pathogenesis of primary myelofibrosis

    Enteric Delivery of Regenerating Family Member 3 alpha Alters the Intestinal Microbiota and Controls Inflammation in Mice With Colitis

    Get PDF
    Background & Aims Paneth cell dysfunction causes deficiencies in intestinal C-type lectins and antimicrobial peptides, which leads to dysbiosis of the intestinal microbiota, alters the mucosal barrier, and promotes development of inflammatory bowel diseases. We investigated whether transgenic (TG) expression of the human regenerating family member 3 alpha gene ( REG3A ) alters the fecal microbiota and affects development of colitis in mice. Methods We performed studies with C57BL/6 mice that express human regenerating family member 3 alpha (hREG3A) in hepatocytes, via the albumin gene promoter. In these mice, hREG3A travels via the bile to the intestinal lumen. Some mice were given dextran sodium sulfate (DSS) to induce colitis. Feces were collected from mice and the composition of the microbiota was analyzed by 16S ribosomal RNA sequencing. The fecal microbiome was also analyzed from mice that express only 1 copy of human REG3A transgene but were fed feces from control mice (not expressing hREG3A) as newborns. Mice expressing hREG3A were monitored for DSS-induced colitis after cohousing or feeding feces from control mice. Colitis was induced in another set of control and hREG3A-TG mice by administration of trinitrobenzene sulfonic acid; some mice were given intrarectal injections of the hREG3A protein. Colon tissues were collected from mice and analyzed by histology and immunohistochemistry to detect mucin 2, as well as by 16S ribosomal RNA fluorescence in situ hybridization, transcriptional analyses, and quantitative polymerase chain reaction. We measured levels of reactive oxygen species (ROS) in bacterial cultures and fecal microbiota using 2′,7′-dichlorofluorescein diacetate and flow cytometry. Results The fecal microbiota of mice that express hREG3A had a significant shift in composition, compared with control mice, with enrichment of Clostridiales (Ruminococcaceae, Lachnospiraceae) and depletion of Bacteroidetes (Prevotellaceae); the TG mice developed less-severe colitis following administration of DSS than control mice, associated with preserved gut barrier integrity and reduced bacterial translocation, epithelial inflammation, and oxidative damage. A similar shift in the composition of the fecal microbiota occurred after a few months in TG mice heterozygous for REG3A that harbored a wild-type maternal microbiota at birth; these mice developed less-severe forms of colitis following DSS administration. Cohoused and germ-free mice fed feces from REG3A- TG mice and given DSS developed less-severe forms of colitis and had reduced lipopolysaccharide activation of the toll-like receptor 4 and increased survival times compared with mice not fed feces from REG3A -TG mice. REG3A TG mice developed only mild colonic inflammation after exposure to 2,4,6-trinitrobenzene sulfonic acid, compared with control mice. Control mice given intrarectal hREG3A and exposed to 2,4,6-trinitrobenzene sulfonic acid showed less colon damage and inflammation than mice not given intrarectal hREG3A. Fecal samples from REG3A- TG mice had lower levels of ROS than feces from control mice during DSS administration. Addition of hREG3A to bacterial cultures reduced levels of ROS and increased survival of oxygen-sensitive commensal bacteria ( Faecalibacterium prausnitzii and Roseburia intestinalis ). Conclusions Mice with hepatocytes that express hREG3A, which travels to the intestinal lumen, are less sensitive to colitis than control mice. We found hREG3A to alter the colonic microbiota by decreasing levels of ROS. Fecal microbiota from REG3A -TG mice protect non-TG mice from induction of colitis. These findings indicate a role for reduction of oxidative stress in preserving the gut microbiota and its ability to prevent inflammation

    Rôle du couple Flt3-ligand/Flt3 et de l'activation des "Mitogen-activated protein kinases" p38 dans la dysmégacaryopoïèse des patients atteints de myélofibrose primitive

    No full text
    The primary myelofibrosis (PMF) is a chronic myeloproliferative neoplasm (NMP) BCR-ABL1-negative associating a dysregulation of hematopoiesis (myeloproliferation, dysmegacaryopoiesis and egress of hematopoietic stem and progenitor cells (HSC / PH)) from an altered bone marrow stroma (osteosclerosis, fibrosis, angiogenesis) to the spleen. The megakaryocyte (MK) is a major player in its pathogenesis through the production of cytokines and fibrotic factors in an inflammatory context. Several arguments suggest that mutations JAK2V617F and MPL515L / K which characterize the NMP are not the initial events of the PMF since they are found only in half of patients. The aim of my work was to investigate whether other abnormalities, genetic or otherwise, could explain the pathogenesis of the PMF. For this, a process parallel to genomics (transcriptome and CGH array), we developed a cell biology approach focused on the role of hematopoietic stroma.Although we have not identified other genomic abnormalities as those described in the literature and in particular, deletion 13q, by genomic approaches we have clarified the limits of this deletion in the PH CD34+ and polymorphonuclear patients. This deletion (chromosomal region 13q14-13q21 minimum) is located 2 megabases (telomeric) of the cluster where is located the FLT gene FLT3. Several arguments have then led to inquire whether the couple was involved in Flt3-ligand/Flt3 deregulation of hematopoiesis, especially in the dysmegakaryopoiesis observed in patients. Among these are: 1) the existence of an expression modulation of genes included in the area of deletion 13q and FLT in the cluster, as gene FLT3 and 2) the fact that Flt3, a key receptor the regulation of primitive hematopoiesis, is often implicated in the pathogenesis of hematologic malignancies and its ligand, Flt3-ligand, was predominantly produced by the hematopoietic stroma.Our study shows dysregulation of Flt3 and p38 MAPKs in CD34+ and PH MK from patients with PMF and this, whatever their Jak2 mutation status. It also shows that persistent stimulation of the axis Flt3/p38 in response to increased production of Flt3 ligand, participates in the dysmegacaryopoiesis that characterizes the disease. Indeed, we have highlighted: 1) an increase in serum Flt3 ligand and its expression by stromal cells and bone marrow and spleen by PH patients with PMF, 2) a specific overexpression of its receptor Flt3 and its phosphorylation in HSC / PH CD34+ and megakaryocytic progenitors (MK), which persist during the MK differentiation, regardless of the mutational status of Jak2 patients, 3) activation of Flt3 in MK progenitors by the Flt3 ligand leads to phosphorylation cascade signaling pathway, p38 MAPK and expression of its target genes such as AP-1, p53, NFATc4, ATF2, IL-8, 4) a restoration of megakaryopoiesis and inhibition of migration (Flt3-ligand)-dependent patients after of MK progenitors by Flt3 or p38 inhibitors.Our results confirm the importance of an alteration of MAPKs in a deregulation of hematopoiesis and highlight the role of a persistent activation of the p38 pathway, via the couple Flt3-ligand/Flt3 in the dysmegakaryopoiesis that characterizes idiopathic myelofibrosis. They also suggest that this dysregulation contributes to the inflammatory process at the origin of the stromal reaction and "bed" of a leukemic transformation potential. The dialogue among impaired hematopoietic cell disease (Bad Seeds), especially the stromal cells and megakaryocyte (Bad Soil), reinforces our concept of "Bad Seeds in Bad Soil". This work could help improve the dialogue with therapeutic approaches targeting the axis Flt3-ligand/Flt3 mediated by activation of p38 which, by reducing the inflammatory process, re-establish a link between the "seed" and the "Soil".La myélofibrose primitive (MFP) est un néoplasme myéloprolifératif (NMP) chronique BCR-ABL1-négatif associant une dérégulation de l’hématopoïèse (myéloprolifération, dysmégacaryopoïèse et migration des cellules souches et progéniteurs hématopoïétiques (CSH/PH)) à une altération du stroma médullaire et splénique (fibrose ostéomyélosclérose, néoangiogenèse). Le mégacaryocyte (MK) est un acteur majeur de sa pathogenèse, via la production de cytokines et facteurs fibrosants, dans un contexte inflammatoire. Plusieurs arguments suggèrent que les mutations JAK2V617F et MPL515L/K qui caractérisent les NMP ne sont pas les événements initiaux de la MFP car elles ne sont retrouvées que chez la moitié des patients. L’objectif de mon travail a été de rechercher si d’autres anomalies, géniques ou non, pouvaient expliquer la pathogenèse de la MFP. Pour cela, parallèlement à une démarche génomique (transcriptome et CGH array), nous avons développé une approche de biologie cellulaire ciblée sur le rôle du stroma hématopoïétique. Bien que n’ayant pas identifié d’autres anomalies génomiques que celles décrites dans la littérature et en particulier, la délétion 13q, les approches génomiques que nous avons développées nous ont permis de préciser les bornes de cette délétion dans les PH CD34+ et les polynucléaires des patients. Cette délétion (région chromosomique minimale 13q14-13q21) est située à 2 mégabases (télomérique) du cluster FLT où est localisé le gène FLT3. Plusieurs arguments nous ont ensuite conduits à rechercher si le couple Flt3-ligand/Flt3 était impliqué dans la dérégulation de l’hématopoïèse et plus particulièrement dans la dysmégacaryopoïèse observée chez les patients. Parmi ceux-ci, citons : 1) l’existence d’une modulation d’expression de gènes inclus dans la zone de délétion 13q et dans le cluster FLT, dont le gène FLT3 et 2) le fait que Flt3, un récepteur clé de la régulation de l’hématopoïèse primitive, soit souvent impliqué dans la pathogenèse d’hémopathies malignes et que son ligand, Flt3-ligand, soit majoritairement produit par le stroma hématopoïétique. Notre étude montre une dérégulation de Flt3 et des MAPKs p38 dans les PH CD34+ et les MK des patients atteints de MFP et ceci, quelque soit leur statut mutationnel Jak2. Elle démontre également que la persistance de la stimulation de l’axe Flt3/p38 en réponse à une production accrue de Flt3 ligand, participe à la dysmégacaryopoïèse qui caractérise la maladie. En effet, nous avons mis en évidence : 1) une augmentation du taux sérique de Flt3 ligand et de son expression par les cellules du stroma médullaire et splénique ainsi que par les PH des patients atteints de MFP, 2) une surexpression spécifique de son récepteur Flt3 et de sa phosphorylation dans les CSH/PH CD34+ et les progéniteurs mégacaryocytaires (MK), qui persistent au cours de la différenciation MK, quelque soit le statut mutationnel de Jak2 des patients, 3) une activation de Flt3 dans les progéniteurs MK en réponse au Flt3 ligand conduisant à la phosphorylation en cascade de la voie de signalisation des MAPKs p38 et à l’expression de ses gènes cibles tels que AP-1, p53, NFATc4, ATF2, IL-8, 4) une restauration de la mégacaryopoïèse et une inhibition de la migration (Flt3-ligand)-dépendante des progéniteurs MK des patients après inhibition de Flt3 ou de p38.Nos résultats confirment l’importance d’une altération des MAPKs dans une dérégulation de l’hématopoïèse et soulignent le rôle d’une activation persistante de la voie p38, via le couple Flt3-ligand/Flt3, dans la dysmégacaryopoïèse qui caractérise la myélofibrose primitive. Ils suggèrent également que cette dérégulation participe au processus inflammatoire à l’origine de la réaction stromale et « lit » d’une transformation leucémique potentielle. Ce dialogue altéré entre les cellules hématopoïétiques pathologiques (Bad seeds), en particulier mégacaryocytaires et les cellules stromales (Bad soil), conforte notre concept « Bad seeds in Bad soil »

    Protein-protein interaction analysis highlights the role of septins in membrane enclosed lumen and mRNA processing

    No full text
    International audienceSeptins are a family of GTP-binding proteins that assemble into non-polar filaments which can be recruited to negatively charged membranes and serve as a scaffold to recruit cytosolic proteins and cytoskeletal elements such as microtubules and actin so that they can perform their important biological functions. Human septins consist of four groups, each with 13 members, and filaments formation usually involve members from each group in specific positions. However, little is known about the molecular mechanisms that drive the binding of septins to membranes and its importance to their biological functions. Here we have built a protein-protein interaction (PPI) network around human septins and highlighted the connections with 170 partners. Functional enrichment by inference of the network of septins and their partners revealed their participation in functions consistent with some of the roles described for septins, including cell cycle, cell division and cell shape, but we also identified septin partners in these functions that had not previously been described. Interestingly, we identified important and multiple connections between septins and mRNA processing and their export from the nucleus. Analysis of the enrichment of gene ontology cellular components highlighted some important interactions between molecules involved in the spliceosome with septin 2 and septin 7 in particular. RNA splicing regulates gene expression, and through it, cell fate, development and physiology. Mutations in components of the in the splicing machinery is linked to several diseases including cancer, thus taken together, the different analyses presented here open new perspectives to elucidate the pathobiological role of septins

    Rôle du couple Flt3-ligand/Flt3 et de l'activation des "Mitogen-activated protein kinases" p38 dans la dysmégacaryopoïèse des patients atteints de myélofibrose primitive.

    No full text
    La myélofibrose primitive (MFP) est un néoplasme myéloprolifératif (NMP) chronique BCR-ABL1-négatif associant une dérégulation de l hématopoïèse (myéloprolifération, dysmégacaryopoïèse et migration des cellules souches et progéniteurs hématopoïétiques (CSH/PH)) à une altération du stroma médullaire et splénique (fibrose ostéomyélosclérose, néoangiogenèse). Le mégacaryocyte (MK) est un acteur majeur de sa pathogenèse, via la production de cytokines et facteurs fibrosants, dans un contexte inflammatoire. Plusieurs arguments suggèrent que les mutations JAK2V617F et MPL515L/K qui caractérisent les NMP ne sont pas les événements initiaux de la MFP car elles ne sont retrouvées que chez la moitié des patients. L objectif de mon travail a été de rechercher si d autres anomalies, géniques ou non, pouvaient expliquer la pathogenèse de la MFP. Pour cela, parallèlement à une démarche génomique (transcriptome et CGH array), nous avons développé une approche de biologie cellulaire ciblée sur le rôle du stroma hématopoïétique. Bien que n ayant pas identifié d autres anomalies génomiques que celles décrites dans la littérature et en particulier, la délétion 13q, les approches génomiques que nous avons développées nous ont permis de préciser les bornes de cette délétion dans les PH CD34+ et les polynucléaires des patients. Cette délétion (région chromosomique minimale 13q14-13q21) est située à 2 mégabases (télomérique) du cluster FLT où est localisé le gène FLT3. Plusieurs arguments nous ont ensuite conduits à rechercher si le couple Flt3-ligand/Flt3 était impliqué dans la dérégulation de l hématopoïèse et plus particulièrement dans la dysmégacaryopoïèse observée chez les patients. Parmi ceux-ci, citons : 1) l existence d une modulation d expression de gènes inclus dans la zone de délétion 13q et dans le cluster FLT, dont le gène FLT3 et 2) le fait que Flt3, un récepteur clé de la régulation de l hématopoïèse primitive, soit souvent impliqué dans la pathogenèse d hémopathies malignes et que son ligand, Flt3-ligand, soit majoritairement produit par le stroma hématopoïétique. Notre étude montre une dérégulation de Flt3 et des MAPKs p38 dans les PH CD34+ et les MK des patients atteints de MFP et ceci, quelque soit leur statut mutationnel Jak2. Elle démontre également que la persistance de la stimulation de l axe Flt3/p38 en réponse à une production accrue de Flt3 ligand, participe à la dysmégacaryopoïèse qui caractérise la maladie. En effet, nous avons mis en évidence : 1) une augmentation du taux sérique de Flt3 ligand et de son expression par les cellules du stroma médullaire et splénique ainsi que par les PH des patients atteints de MFP, 2) une surexpression spécifique de son récepteur Flt3 et de sa phosphorylation dans les CSH/PH CD34+ et les progéniteurs mégacaryocytaires (MK), qui persistent au cours de la différenciation MK, quelque soit le statut mutationnel de Jak2 des patients, 3) une activation de Flt3 dans les progéniteurs MK en réponse au Flt3 ligand conduisant à la phosphorylation en cascade de la voie de signalisation des MAPKs p38 et à l expression de ses gènes cibles tels que AP-1, p53, NFATc4, ATF2, IL-8, 4) une restauration de la mégacaryopoïèse et une inhibition de la migration (Flt3-ligand)-dépendante des progéniteurs MK des patients après inhibition de Flt3 ou de p38.Nos résultats confirment l importance d une altération des MAPKs dans une dérégulation de l hématopoïèse et soulignent le rôle d une activation persistante de la voie p38, via le couple Flt3-ligand/Flt3, dans la dysmégacaryopoïèse qui caractérise la myélofibrose primitive. Ils suggèrent également que cette dérégulation participe au processus inflammatoire à l origine de la réaction stromale et lit d une transformation leucémique potentielle. Ce dialogue altéré entre les cellules hématopoïétiques pathologiques (Bad seeds), en particulier mégacaryocytaires et les cellules stromales (Bad soil), conforte notre concept Bad seeds in Bad soil . Ce travail pourrait contribuer à l amélioration de ce dialogue par des approches thérapeutiques ciblées sur l axe Flt3-ligand/Flt3 médié par l activation de p38 qui, en réduisant le processus inflammatoire, rétablirait un lien entre le Seed et le Soil .The primary myelofibrosis (PMF) is a chronic myeloproliferative neoplasm (NMP) BCR-ABL1-negative associating a dysregulation of hematopoiesis (myeloproliferation, dysmegacaryopoiesis and egress of hematopoietic stem and progenitor cells (HSC / PH)) from an altered bone marrow stroma (osteosclerosis, fibrosis, angiogenesis) to the spleen. The megakaryocyte (MK) is a major player in its pathogenesis through the production of cytokines and fibrotic factors in an inflammatory context. Several arguments suggest that mutations JAK2V617F and MPL515L / K which characterize the NMP are not the initial events of the PMF since they are found only in half of patients. The aim of my work was to investigate whether other abnormalities, genetic or otherwise, could explain the pathogenesis of the PMF. For this, a process parallel to genomics (transcriptome and CGH array), we developed a cell biology approach focused on the role of hematopoietic stroma.Although we have not identified other genomic abnormalities as those described in the literature and in particular, deletion 13q, by genomic approaches we have clarified the limits of this deletion in the PH CD34+ and polymorphonuclear patients. This deletion (chromosomal region 13q14-13q21 minimum) is located 2 megabases (telomeric) of the cluster where is located the FLT gene FLT3. Several arguments have then led to inquire whether the couple was involved in Flt3-ligand/Flt3 deregulation of hematopoiesis, especially in the dysmegakaryopoiesis observed in patients. Among these are: 1) the existence of an expression modulation of genes included in the area of deletion 13q and FLT in the cluster, as gene FLT3 and 2) the fact that Flt3, a key receptor the regulation of primitive hematopoiesis, is often implicated in the pathogenesis of hematologic malignancies and its ligand, Flt3-ligand, was predominantly produced by the hematopoietic stroma.Our study shows dysregulation of Flt3 and p38 MAPKs in CD34+ and PH MK from patients with PMF and this, whatever their Jak2 mutation status. It also shows that persistent stimulation of the axis Flt3/p38 in response to increased production of Flt3 ligand, participates in the dysmegacaryopoiesis that characterizes the disease. Indeed, we have highlighted: 1) an increase in serum Flt3 ligand and its expression by stromal cells and bone marrow and spleen by PH patients with PMF, 2) a specific overexpression of its receptor Flt3 and its phosphorylation in HSC / PH CD34+ and megakaryocytic progenitors (MK), which persist during the MK differentiation, regardless of the mutational status of Jak2 patients, 3) activation of Flt3 in MK progenitors by the Flt3 ligand leads to phosphorylation cascade signaling pathway, p38 MAPK and expression of its target genes such as AP-1, p53, NFATc4, ATF2, IL-8, 4) a restoration of megakaryopoiesis and inhibition of migration (Flt3-ligand)-dependent patients after of MK progenitors by Flt3 or p38 inhibitors.Our results confirm the importance of an alteration of MAPKs in a deregulation of hematopoiesis and highlight the role of a persistent activation of the p38 pathway, via the couple Flt3-ligand/Flt3 in the dysmegakaryopoiesis that characterizes idiopathic myelofibrosis. They also suggest that this dysregulation contributes to the inflammatory process at the origin of the stromal reaction and "bed" of a leukemic transformation potential. The dialogue among impaired hematopoietic cell disease (Bad Seeds), especially the stromal cells and megakaryocyte (Bad Soil), reinforces our concept of "Bad Seeds in Bad Soil". This work could help improve the dialogue with therapeutic approaches targeting the axis Flt3-ligand/Flt3 mediated by activation of p38 which, by reducing the inflammatory process, re-establish a link between the "seed" and the "Soil".PARIS11-SCD-Bib. électronique (914719901) / SudocSudocFranceF

    Impact of HCV Infection on Hepatocyte Polarity and Plasticity

    No full text
    The hepatitis C virus (HCV) is an oncogenic virus that alters the cell polarization machinery in order to enter the hepatocyte and replicate. While these alterations are relatively well defined, their consequences in the evolution of the disease remain poorly documented. Since 2012, HCV infection can be effectively cured with the advent of direct acting antivirals (DAA). Nevertheless, patients cured of their HCV infection still have a high risk of developing hepatocellular carcinoma (HCC). Importantly, it has been shown that some of the deregulations induced by HCV are maintained despite a sustained virologic response (SVR), including the down-regulation of some hepatocyte functions such as bile acid metabolism, exemplifying cell dedifferentiation, and the up-regulation of the epithelial–mesenchymal transition (EMT). EMT is a process by which epithelial cells lose their differentiation and their specific polarity to acquire mesenchymal cell properties, including migration and extracellular matrix remodeling capabilities. Of note, epithelial cell polarity acts as a gatekeeper against EMT. Thus, it remains important to elucidate the mechanisms by which HCV alters polarity and promotes EMT that could participate in viral-induced hepatic carcinogenesis. In this review, we define the main steps involved in the polarization process of epithelial cells and recall the essential cellular actors involved. We also highlight the particularities of hepatocyte polarity, responsible for their unique morphology. We then focus on the alterations by HCV of epithelial cell polarity and the consequences of the transformation of hepatocytes involved in the carcinogenesis process
    • …
    corecore